Skip to main content
Log in

On parameter estimation in an in vitro compartmental model for drug-induced enzyme production in pharmacotherapy

  • Published:
Applications of Mathematics Aims and scope Submit manuscript

Abstract

A pharmacodynamic model introduced earlier in the literature for in silico prediction of rifampicin-induced CYP3A4 enzyme production is described and some aspects of the involved curve-fitting based parameter estimation are discussed. Validation with our own laboratory data shows that the quality of the fit is particularly sensitive with respect to an unknown parameter representing the concentration of the nuclear receptor PXR (pregnane X receptor). A detailed analysis of the influence of that parameter on the solution of the model’s system of ordinary differential equations is given and it is pointed out that some ingredients of the analysis might be useful for more general pharmacodynamic models. Numerical experiments are presented to illustrate the performance of related parameter estimation procedures based on least-squares minimization.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Similar content being viewed by others

References

  1. D. Z. D’Argenio, A. Schumitzky, X. Wang: ADAPT 5 User’s Guide: Pharmacokinetic/Pharmacodynamic Systems Analysis Software. Biomedical Simulations Resource, Los Angeles, 2009, Available at https://doi.org/bmsr.usc.edu/software/adapt/users-guide/.

    Google Scholar 

  2. S. Dhillon, A. Kostrzewski, (eds.): Clinical Pharmacokinetics. Pharmaceutical Press, London, 2006.

    Google Scholar 

  3. J. Duintjer Tebbens, M. Azar, E. Friedmann, M. Lanzendörfer, P. Pávek: Mathematical models in the description of pregnane X receptor (PXR)-regulated cytochrome P450 enzyme induction. Int. J. Mol. Sci. 19(2018), 1785.

    Article  Google Scholar 

  4. A. Funahashi, M. Morohashi, H. Kitano, N. Tanimura: CellDesigner: a process diagram editor for gene-regulatory and biochemical networks. BIOSILICO 1(2003), 159–162.

    Article  Google Scholar 

  5. The GNU Fortran compiler. Available at https://doi.org/gcc.gnu.org/fortran/.

  6. A. C. Hindmarsh: Large ordinary differential equation systems and software. Control Systems Magazine 2 (1982), 24–30.

    Article  Google Scholar 

  7. A. C. Hindmarsh: ODEPACK, a systematized collection of ODE solvers. Scientific Computing 1982 (R. S. Stepleman et al., eds.). IMACS Transactions on Scientific Computation I, North-Holland Publishing, Amsterdam, 1983, pp. 55–64.

    Google Scholar 

  8. H. M. Jones, K. Rowland-Yeo: Basic Concepts in Physiologically Based Pharmacokinetic Modeling in Drug Discovery and Development. CPT: Pharmacometrics & Systems Pharmacology 2 (2013), Article ID e63, 12 pages.

    Google Scholar 

  9. N. S. Luke, M. J. DeVito, I. Shah, H. A. El-Masri: Development of a quantitative model of pregnane X receptor (PXR) mediated xenobiotic metabolizing enzyme induction. Bull. Math. Biol. 72 (2010), 1799–1819.

    Article  MathSciNet  MATH  Google Scholar 

  10. L. Lukšan, M. Tůma, C. Matonoha, J. Vlček, N. Ramešová, M. Šiška, J. Hartman: UFO 2017. Interactive System for Universal Functional Optimization. Technical Report V-1252, Institute for Computer Science CAS, Praha, 2017. Available at https://doi.org/www.cs.cas.cz/luksan/ufo.html.

    Google Scholar 

  11. D. J. Lunn, N. Best, A. Thomas, J. Wakefield, D. J. Spiegelhalter: Bayesian analysis of population PK/PD models: General concepts and software. J. Pharmacokinetics Pharmacodynamics 29 (2002), 271–307.

    Article  Google Scholar 

  12. MATLAB. Mathworks, Inc., 2018. Available at https://doi.org/www.mathworks.com/products/matlab.html.

  13. C. Moler, C. Van Loan: Nineteen dubious ways to compute the exponential of a matrix, twenty-five years later. SIAM Rev. 45 (2003), 3–49.

    Article  MathSciNet  MATH  Google Scholar 

  14. J. A. Nelder, R. Mead: A simplex method for function minimization. Computer J. 4 (1965), 308–313.

    Article  MathSciNet  MATH  Google Scholar 

  15. NONMEM 7.3. ICON, Inc., 1990–2016. Available at https://doi.org/www.iconplc.com/innovation/nonmem/.

  16. L. Petzold: Automatic selection of methods for solving stiff and nonstiff systems of ordinary diferential equations. SIAM J. Sci. Stat. Comput. 4 (1983), 136–148.

    Article  MATH  Google Scholar 

  17. L. Shargel, A. B. C. Yu: Applied Biopharmaceutics & Pharmacokinetics. McGraw-Hill Education, New York, 2016.

    Google Scholar 

  18. Simcyp simulator. Certara, 2012. Available at https://doi.org/www.certara.com/software/.

  19. J. W. Spruill, W. E. Wade, T. J. DiPiro, A. R. Blouin, M. J. Pruemer: Concepts in Clinical Pharmacokinetics. American Society of Health-System Pharmacists, Bethesda, 2014.

    Google Scholar 

  20. P. Zhao, M. Rowland, S.-M. Huang: Best practice in the use of physiologically based pharmacokinetic modeling and simulation to address clinical pharmacology regulatory questions. Clinical Pharmacology & Therapeutics 92 (2012), 17–20.

    Article  Google Scholar 

  21. Z. Zheng, P. S. Stewart: Penetration of rifampin through staphylococcus epidermidis biofilms. Antimicrob. Agents Chemoter 46 (2002), 900–903.

    Article  Google Scholar 

Download references

Acknowledgement

We would like to thank Prof. Petr Pávek for the laboratory experiments described in this manuscript; they were performed under his supervision and in his laboratory at the Faculty of Pharmacy of Charles university in Hradec Králové.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Jurjen Duintjer Tebbens, Ctirad Matonoha, Andreas Matthios or Štěpán Papáček.

Additional information

The work of Jurjen Duintjer Tebbens and Ctirad Matonoha was supported by the long-term strategic development financing of the Institute of Computer Science (RVO: 67985807) of the Czech Academy of Sciences. The work of Jurjen Duintjer Tebbens was also supported by the Czech science funding agency (GACR 17-06841S). The work of Andreas Matthios was supported by the grant agency of Charles University project GAUK 110/50/85003 and by SVV 260 414. The work of Štěpán Papáček was supported by the Ministry of Education, Youth and Sports of the Czech Republic — projects “CENAKVA” (No. CZ.1.05/2.1.00/01.0024), “CENAKVA II” (No. LO1205 under the NPU I program) and The CENAKVA Centre Development (No. CZ.1.05/2.1.00/19.0380)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Duintjer Tebbens, J., Matonoha, C., Matthios, A. et al. On parameter estimation in an in vitro compartmental model for drug-induced enzyme production in pharmacotherapy. Appl Math 64, 253–277 (2019). https://doi.org/10.21136/AM.2019.0284-18

Download citation

  • Received:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.21136/AM.2019.0284-18

Keywords

MSC 2010

Navigation